Human diseases

人类疾病
  • 文章类型: Journal Article
    N6-甲基腺苷(m6A)是真核生物中最丰富的转录后动态RNA修饰过程,广泛参与细胞生长,胚胎发育和免疫稳态。m6A最深刻的生物学功能之一是调节RNA代谢,从而决定了RNA的命运。值得注意的是,m6A介导的有组织的RNA代谢的调节关键依赖于无膜细胞器(MLO)在细胞核和细胞质中的组装,比如核斑点,应力颗粒和加工体。此外,m6A相关的MLO在控制包括转录在内的各种RNA代谢过程中发挥关键作用,拼接,运输,衰减和翻译。然而,新出现的证据表明,m6A水平失调有助于在一系列人类疾病中形成病理性凝聚物,包括肿瘤发生,生殖疾病,神经系统疾病和呼吸系统疾病。迄今为止,m6A调节与RNA代谢相关的生物分子缩合物聚集的分子机制尚不清楚.在这次审查中,我们全面总结了与M6A相关的MLO的最新生化过程,特别关注它们对RNA代谢的影响以及它们在疾病发展和相关生物学机制中的关键作用。此外,我们认为,m6A相关的MLO可作为疾病进展的预测标志物和未来潜在的药物靶点.
    N6-methyladenosine (m6A) is the most abundant post-transcriptional dynamic RNA modification process in eukaryotes, extensively implicated in cellular growth, embryonic development and immune homeostasis. One of the most profound biological functions of m6A is to regulate RNA metabolism, thereby determining the fate of RNA. Notably, the regulation of m6A-mediated organized RNA metabolism critically relies on the assembly of membraneless organelles (MLOs) in both the nucleus and cytoplasm, such as nuclear speckles, stress granules and processing bodies. In addition, m6A-associated MLOs exert a pivotal role in governing diverse RNA metabolic processes encompassing transcription, splicing, transport, decay and translation. However, emerging evidence suggests that dysregulated m6A levels contribute to the formation of pathological condensates in a range of human diseases, including tumorigenesis, reproductive diseases, neurological diseases and respiratory diseases. To date, the molecular mechanism by which m6A regulates the aggregation of biomolecular condensates associated with RNA metabolism is unclear. In this review, we comprehensively summarize the updated biochemical processes of m6A-associated MLOs, particularly focusing on their impact on RNA metabolism and their pivotal role in disease development and related biological mechanisms. Furthermore, we propose that m6A-associated MLOs could serve as predictive markers for disease progression and potential drug targets in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基因治疗近年来取得了实质性进展,成为治疗各种人类疾病的建设性策略。这篇综述全面概述了这些发展,专注于它们在不同疾病环境中的不同应用。它探索了基因传递系统的进化,包括病毒(如腺相关病毒;AAV)和非病毒方法,并评估其固有的优势和局限性。此外,该综述深入研究了在靶向特定组织和细胞类型方面取得的进展,跨越眼睛,肝脏,肌肉,和中枢神经系统,其中,利用这些基因技术。这种有针对性的方法对于解决广泛的遗传疾病至关重要,如遗传性溶酶体贮积病,神经退行性疾病,和心血管疾病。最近的临床试验和基因治疗的成功结果,特别是那些涉及AAV和成簇的规则间隔短回文重复(CRISPR)-CRISPR相关蛋白,被突出显示,阐明这种方法在疾病治疗中的转化潜力。综述了基因治疗的现状,其前景,及其显著改善患者预后和生活质量的能力。通过提供全面的分析,这篇综述为研究人员提供了宝贵的见解,临床医生,和利益相关者,丰富了正在进行的关于疾病治疗轨迹的话语。
    Gene therapy has witnessed substantial advancements in recent years, becoming a constructive tactic for treating various human diseases. This review presents a comprehensive overview of these developments, with a focus on their diverse applications in different disease contexts. It explores the evolution of gene delivery systems, encompassing viral (like adeno-associated virus; AAV) and nonviral approaches, and evaluates their inherent strengths and limitations. Moreover, the review delves into the progress made in targeting specific tissues and cell types, spanning the eye, liver, muscles, and central nervous system, among others, using these gene technologies. This targeted approach is crucial in addressing a broad spectrum of genetic disorders, such as inherited lysosomal storage diseases, neurodegenerative disorders, and cardiovascular diseases. Recent clinical trials and successful outcomes in gene therapy, particularly those involving AAV and the clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated proteins, are highlighted, illuminating the transformative potentials of this approach in disease treatment. The review summarizes the current status of gene therapy, its prospects, and its capacity to significantly ameliorate patient outcomes and quality of life. By offering comprehensive analysis, this review provides invaluable insights for researchers, clinicians, and stakeholders, enriching the ongoing discourse on the trajectory of disease treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基因编辑是一种不断发展的基因工程技术,它可以对广谱的基因调节疾病进行精确编辑以实现治愈性治疗,也有可能作为疾病常规治疗的辅助手段。基因编辑技术,主要基于成簇的规则间隔回文重复(CRISPR)-CRISPR相关蛋白系统,能够在体细胞中产生遗传修饰,为广泛的人类疾病的基因治疗提供了有希望的新策略。目前,基因编辑技术在人类多种疾病中显示出巨大的应用前景,不仅在治疗潜力方面,而且在人类疾病动物模型的构建方面。本文阐述了基因编辑技术在血液病中的应用,实体瘤,免疫疾病,眼科疾病,和代谢性疾病;重点介绍了基因编辑技术在镰状细胞病中的治疗策略;概述了基因编辑技术在人类疾病动物模型构建中的作用;并讨论了基因编辑技术在疾病治疗中的局限性,旨在为基因编辑技术在人类疾病中的应用提供重要参考。
    Gene editing is a growing gene engineering technique that allows accurate editing of a broad spectrum of gene-regulated diseases to achieve curative treatment and also has the potential to be used as an adjunct to the conventional treatment of diseases. Gene editing technology, mainly based on clustered regularly interspaced palindromic repeats (CRISPR)-CRISPR-associated protein systems, which is capable of generating genetic modifications in somatic cells, provides a promising new strategy for gene therapy for a wide range of human diseases. Currently, gene editing technology shows great application prospects in a variety of human diseases, not only in therapeutic potential but also in the construction of animal models of human diseases. This paper describes the application of gene editing technology in hematological diseases, solid tumors, immune disorders, ophthalmological diseases, and metabolic diseases; focuses on the therapeutic strategies of gene editing technology in sickle cell disease; provides an overview of the role of gene editing technology in the construction of animal models of human diseases; and discusses the limitations of gene editing technology in the treatment of diseases, which is intended to provide an important reference for the applications of gene editing technology in the human disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    人类肠道微生物群,由数万亿的微生物组成,编码影响宿主生理学许多方面的不同代谢途径。肠道细菌与宿主相互作用的一个关键方式是通过产生小的代谢物。这些微生物群依赖的代谢物中的几种,比如短链脂肪酸,已被证明可以调节宿主疾病。在这次审查中,我们研究了如何使用代谢组学平台识别疾病相关的代谢特征,代谢组学应用于肠道微生物群-疾病相互作用。我们进一步探索宏基因组和代谢组学数据在人体研究中的整合如何促进精准医学中的生物标志物发现。
    The human gut microbiota, consisting of trillions of microorganisms, encodes diverse metabolic pathways that impact numerous aspects of host physiology. One key way in which gut bacteria interact with the host is through the production of small metabolites. Several of these microbiota-dependent metabolites, such as short-chain fatty acids, have been shown to modulate host diseases. In this review, we examine how disease-associated metabolic signatures are identified using metabolomic platforms, and where metabolomics is applied in gut microbiota-disease interactions. We further explore how integration of metagenomic and metabolomic data in human studies can facilitate biomarkers discoveries in precision medicine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在医学科学中使用的大多数众所周知的药物基因组学研究有助于我们对药物相互作用的理解。它对治疗和药物开发有重大影响。药物基因组学的广泛应用是治疗进展所必需的。主要关注的是基因和复杂的基因系统如何影响身体对药物的反应。作为临床治疗领域的最新发展的结果,已经发现了帮助识别或多或少可能响应于某种药物的患者组的新型生物标志物。它旨在通过在正确的时间以正确的剂量给予适当的药物来改善定制治疗,并确保发出正确的处方。遗传的结合,环境,影响药物药代动力学和/或药效学的患者变量导致药物反应的个体间差异。药物开发,疾病易感性,和治疗效果都受到药物基因组学的影响。这项工作的目的是给出一个评论,可能作为创建新的药物基因组学应用的基础,技术,或策略。
    The majority of the well-known pharmacogenomics research used in the medical sciences contributes to our understanding of medication interactions. It has a significant impact on treatment and drug development. The broad use of pharmacogenomics is required for the progress of therapy. The main focus is on how genes and an intricate gene system affect the body\'s reaction to medications. Novel biomarkers that help identify a patient group that is more or less likely to respond to a certain medication have been discovered as a result of recent developments in the field of clinical therapeutics. It aims to improve customized therapy by giving the appropriate drug at the right dose at the right time and making sure that the right prescriptions are issued. A combination of genetic, environmental, and patient variables that impact the pharmacokinetics and/or pharmacodynamics of medications results in interindividual variance in drug response. Drug development, illness susceptibility, and treatment efficacy are all impacted by pharmacogenomics. The purpose of this work is to give a review that might serve as a foundation for the creation of new pharmacogenomics applications, techniques, or strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    亲环蛋白(Cyps),表征为肽基丙氨酰基顺反异构酶(PPI酶),高度保守和无处不在,在蛋白质折叠和细胞信号传导中起着至关重要的作用。本文综述了Cyps介导的生化通路,包括它们参与病理状态,如病毒复制,炎症,和癌症进展,强调Cyp抑制的治疗潜力。本文综述了Cyp抑制剂(CypI)的研究进展,特别是非免疫抑制环孢素A(CsA)衍生物,突出了它们作为治疗剂的重要性。检查了CsA衍生物的结构和功能细微差别,包括它们的功效,作用机制,以及治疗益处和脱靶效应之间的平衡。对CypI的景观进行了评估,以强调临床上需要有针对性的方法来利用Cyps的复杂生物学,并提出了未来的研究方向,这些研究方向可能会增强非免疫抑制性CsA衍生物在治疗Cyps发挥关键病理作用的疾病中的效用。
    Cyclophilins (Cyps), characterized as peptidyl-prolyl cis-trans isomerases (PPIases), are highly conserved and ubiquitous, playing a crucial role in protein folding and cellular signaling. This review summarizes the biochemical pathways mediated by Cyps, including their involvement in pathological states such as viral replication, inflammation, and cancer progression, to underscore the therapeutic potential of Cyp inhibition. The exploration of Cyp inhibitors (CypI) in this review, particularly non-immunosuppressive cyclosporine A (CsA) derivatives, highlights their significance as therapeutic agents. The structural and functional nuances of CsA derivatives are examined, including their efficacy, mechanism of action, and the balance between therapeutic benefits and off-target effects. The landscape of CypI is evaluated to emphasize the clinical need for targeted approaches to exploit the complex biology of Cyps and to propose future directions for research that may enhance the utility of non-immunosuppressive CsA derivatives in treating diseases where Cyps play a key pathological role.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    作为表观遗传机制的一部分,微RNA(miRNA)被真核生物广泛利用。通过多种方式调节基因表达,这些短的RNA介导关键的生理过程。这表明miRNA生物发生和表达的异常可以追溯到多种疾病。此外,miRNA是有希望的临床候选,尤其是临床前诊断.miRNA的Let家族是最早被发现的家族之一。作为这个类别的重要成员,对Let-7e进行了广泛的研究。绝大多数证据表明let-7e失调与疾病的发生和进展之间存在关联,包括恶性肿瘤.因为它们的效果取决于疾病的遗传特征和受影响的组织,不同的miRNA在各种疾病中发挥不同的作用。然而,在miRNA研究中最重要的是,只有一个miRNA可以在确切的时间靶向细胞中的许多mRNA,因此总结单一miRNA在人类疾病中的作用可以为疾病的检测和治疗提供更好的见解。这项研究的目的是更深入地了解let-7e如何在人体细胞中发挥作用,以便在临床诊断中更有效地利用它。预后,和治疗。我们回顾了关于let-7e的研究,关注由这种miRNA控制的生物过程的分子基础,这些过程有助于许多疾病的发展和病因。
    As part of the epigenetic machinery, microRNAs (miRNAs) are extensively utilized by eukaryotes. By modulating gene expression in a variety of ways, these short RNAs mediate crucial physiological processes. This suggests that abnormalities in miRNA biogenesis and expression can be traced back to a variety of diseases. In addition, miRNAs are promising clinical candidates, especially for preclinical diagnosis. The Let family of miRNAs was one of the first to be discovered. As a prominent member of this category, extensive research has been conducted on Let-7e. The vast majority of evidence indicates an association between let-7e dysregulation and the onset and progression of disease, including malignancies. Because their effect depends on the genetic profile of disease and the affected tissue, different miRNAs play diverse roles in various diseases. However, what counts in miRNA studies is that just one miRNA may target numerous mRNAs in a cell at the exact time, therefore summarizing the effect of a single miRNA in human diseases can provide better insights into disease detection and treatment. The goal of this study is to gain a deeper understanding of how let-7e functions in human cells so that it can be utilized more effectively in clinical settings for diagnosis, prognosis, and treatment. We have reviewed the research on let-7e, focusing on the molecular underpinnings of biological processes controlled by this miRNA that contribute to the development and etiology of numerous disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    自从古代观察自然节律以来,时间生物学领域取得了显着进步。生物钟复杂的分子结构,它们在哺乳动物体内的等级组织,它们在器官生理学中的关键作用突出了这些内部计时机制的复杂性和重要性。在人类中,昼夜节律表型在个体之间和整个个体的寿命中表现出相当大的变异性。人类时间生物学的机械研究中的基本挑战来自于从大多数器官进行连续采样的困难。体外研究生物钟的概念依赖于UeliSchibler及其同事的开创性发现,即体内几乎每个细胞都有自主分子振荡器。昼夜节律生物发光记者的出现为这种方法提供了新的视角,能够对培养细胞中的细胞自主时钟进行高分辨率连续测量,跟随体外同步脉冲。StevenA.Brown的工作提供了令人信服的证据,表明在体外培养的原代小鼠和人皮肤成纤维细胞中评估的时钟特征代表了体内内部时钟特性的可靠估计。体外方法研究培养的外植体和原代细胞中的分子人钟,由史蒂夫·布朗开创,代表了一个宝贵的工具,用于评估昼夜节律特征的个体差异以及综合遗传,生化和功能分析。在更广泛的背景下,这种可靠和微创的方法提供了一个独特的视角,用于解开在生理环境和各种病理中几乎任何人体组织中操作的振荡器的功能输入和输出。
    The field of chronobiology has advanced significantly since ancient observations of natural rhythms. The intricate molecular architecture of circadian clocks, their hierarchical organization within the mammalian body, and their pivotal roles in organ physiology highlight the complexity and significance of these internal timekeeping mechanisms. In humans, circadian phenotypes exhibit considerable variability among individuals and throughout the individual\'s lifespan. A fundamental challenge in mechanistic studies of human chronobiology arises from the difficulty of conducting serial sampling from most organs. The concept of studying circadian clocks in vitro relies on the groundbreaking discovery by Ueli Schibler and colleagues that nearly every cell in the body harbours autonomous molecular oscillators. The advent of circadian bioluminescent reporters has provided a new perspective for this approach, enabling high-resolution continuous measurements of cell-autonomous clocks in cultured cells, following in vitro synchronization pulse. The work by Steven A. Brown has provided compelling evidence that clock characteristics assessed in primary mouse and human skin fibroblasts cultured in vitro represent a reliable estimation of internal clock properties in vivo. The in vitro approach for studying molecular human clocks in cultured explants and primary cells, pioneered by Steve Brown, represents an invaluable tool for assessing inter-individual differences in circadian characteristics alongside comprehensive genetic, biochemical and functional analyses. In a broader context, this reliable and minimally invasive approach offers a unique perspective for unravelling the functional inputs and outputs of oscillators operative in nearly any human tissue in physiological contexts and across various pathologies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    介体复合物是RNA聚合酶II转录的必需共调节因子。最近的发展表明,介体功能是转录调控之间的联系,基因组组织和DNA修复机制,包括核苷酸切除修复,基底切除修复,和同源重组。这些过程的功能障碍通常与人类病理有关,越来越多的证据表明Mediator参与了癌症,神经学,代谢和传染病。介体功能的分子机制的详细破译,在不同的生物模型中使用跨学科的方法,并考虑到这个复合体的所有功能,将有助于我们对相关人类疾病的理解。
    The Mediator complex is an essential coregulator of RNA polymerase II transcription. More recent developments suggest Mediator functions as a link between transcription regulation, genome organisation and DNA repair mechanisms including nucleotide excision repair, base excision repair, and homologous recombination. Dysfunctions of these processes are frequently associated with human pathologies, and growing evidence shows Mediator involvement in cancers, neurological, metabolic and infectious diseases. The detailed deciphering of molecular mechanisms of Mediator functions, using interdisciplinary approaches in different biological models and considering all functions of this complex, will contribute to our understanding of relevant human diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    小的非编码RNA(sncRNAs),如microRNAs(miRNAs),小干扰RNA(siRNA),PIWI相互作用RNA(piRNAs),和转移RNA(tRNA)衍生的小RNA(tsRNAs),在调节各种细胞和发育过程中发挥重要作用。在过去的三十年里,研究人员已经从各种生物体中发现了新的sncRNA种类。这些分子表现出动态表达和不同的功能,在健康和疾病状态下,它们都会通过RNA修饰受到复杂的调节。值得注意的是,配子中的某些sncRNAs,尤其是精子,响应环境刺激并促进表观遗传。总的来说,对sncRNA功能和机制的深入理解加速了基于小RNA的疗法的发展。在这次审查中,我们介绍了该领域的最新进展,包括新的sncRNA种类和RNA修饰的调节影响。我们还讨论了当前与使用小RNA作为生物标志物或治疗药物相关的限制和挑战。
    Small non-coding RNAs (sncRNAs), such as microRNAs (miRNAs), small interfering RNAs (siRNAs), PIWI-interacting RNAs (piRNAs), and transfer RNA (tRNA)-derived small RNAs (tsRNAs), play essential roles in regulating various cellular and developmental processes. Over the past three decades, researchers have identified novel sncRNA species from various organisms. These molecules demonstrate dynamic expression and diverse functions, and they are subject to intricate regulation through RNA modifications in both healthy and diseased states. Notably, certain sncRNAs in gametes, particularly sperm, respond to environmental stimuli and facilitate epigenetic inheritance. Collectively, the in-depth understanding of sncRNA functions and mechanisms has accelerated the development of small RNA-based therapeutics. In this review, we present the recent advances in the field, including new sncRNA species and the regulatory influences of RNA modifications. We also discuss the current limitations and challenges associated with using small RNAs as either biomarkers or therapeutic drugs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号